throbber
XENOTRANSPLAMATION II
`
`Abstract# 845
`Poster Board #-Session: P232-I1
`ROLE OF NITRIC OXIDE AND ENDOTHELIN IN ISCHEMIA/
`REPERFUSION INJURY IN THE RAT PANCREAS. NicholasT. Stowe,l
`AM V. Robinson,l Mike S. Simonson? James A. Schulak.'.' 'Surgery, Case
`Western Reserve University, Cleveland, OH; 21nternal Medicine, Case
`Western Reserve University. Cleveland, OH: *Surgery. University Hospitals,
`Cleveland, OH.
`Both nitnc oxide (NO) and endothelin (ET-I) control local vascular resistance and
`have been implicated in ischemidreperfusion (I/R) injury in various organs. The
`purpose of this study was to assess the role of NO and ET-I following I/R injury
`in the pancreas by infusing the NO precursor I-arginine. In anesthetized Sprague
`Dawley rats, the pancreas was isolated through a midline incision. Pancreatic blood
`flow (PBF) was measured in the inferior splenic artery with an ultrasonic flow
`probe before and 90 min after ischemia. Two groups were studied. In the control
`ischemia group, the pancreas was subjected only to 60 min of ischemia by occluding
`the inferior splenic artery. In the treated ischemia group, the NO precursor, 1-
`arginine (4Oomghg. i.v.). was infused for 30 min pnor to ischemia. At the end of the
`experiments, blood samples were taken for amylase and lipase activity. In selected
`experiments, plasma ET-I levels and immunohistochemical analysis of ET-I were
`done. In
`the untreated
`ischemia group (n=4).
`ischemia caused a
`7X.lfl.9%(meamtSEM) decrease in PBF from 0.39M.04 mVmin to 0.07M.01 ml/
`min. In contrast, rats pretreated with I-arginine (n=3) experienced only a 41 .%10.6%
`drop in PBF from 0.76M.02 ml/min to 0.46M.05 ml/min, a value significantly
`different (p4.05) from the untreated controls. Lipase values were significantly
`lower after ischemia in the I-arginine treated group at 34.5f11.5 U/L (p<O.05)
`compared to the untreated ischemic group. Amylase values were also lower at
`2688f530 U/L (p<O.lO). In a separate group of experiments ( n d ) . plasma ET-I
`levels were significantly elevated following 60 min of ischemia (1.17M.12 vs
`0.66M.15 pg/ml, in controls, p<O 05). lmmunohistochemical localiiation of ET-I
`peptide following ischemia exhibited increased staining in the vascular endothelium.
`I/R injury in the pancreas is characterized by a decrease in PBF, an increase in
`amylase and lipase activity and an increase in plasma ET-I levels. Pretreatment
`with I-arginine significantly ameliorated the drop in PBF and decreased the levels of
`lipase and amylaw following ischemia. Use of the NO precursor. I-arginine. can
`enhance the production of NO which could antagonize the vasoconstrictor effects
`of ET-I. Such an approach may be useful in the transplantation setting to minimize
`I/R injury and preserve graft function.
`
`POSTER SESSION 11:
`I1
`XENOTRANSPLANTATION
`
`Abstract# 846
`Poster Board #-Session: P233-I1
`LYTRAGRAFl' EXPRESSION OF PROTECllON GEhES HO-1 Ah'D Bd-
`2 IN PRIiMARY HEART XENOGRAFTS: A LACK OF PROTECTION
`EF%E4TACAINSTXENOGRAFTHYPERACWI'EREJE(JT1ONNUCED
`BY HYPER-IMMUNE SERA. Gordon D. Wu. Yang-Sung Jin. Vaughn A.
`Starnes, Donald V. Cramer. 'Cardiothoracic Surgery. Keck School of
`Medicine, USC. Los Angeles, CA.
`Heme-oxygenase-l (HO-1) and Bcl genes are expressed by long-surviving rodent
`xenografts and have been suggested to have a protective role in accommodating
`xenografts against antibody InJUQ!. Expression of these genes by organ xenografts in
`early stages of humoral rejection has not been carefully examined. We have recently
`conducted series of studies on the hamster-to-rat heart transplant model to see
`whether intragraft expression of protection genes is associated with protecting
`xenograft from antibody rejection. First, we \tudied intragraft expression of HO-1
`and Bcl-2 genes at different time points (0.60 minutes, 12,24,48,72 and 96 hours)
`post-transplantation (PTx). All the xenografl ramples (n=3 in each Group) were
`snap-frozen, cryostat sectioned and subjected to immunohistochemistry using
`antibodies against HO-I. Bcl-2, and Bax. We found that hamster heart tissue with
`cold preservation (Group TO) expressed strong HO-I protein. and heart xenografts
`in other groups exhibited moderate HO-I staining. lmmunostain for Bcl-2 was also
`positive in all of the xenografts except Group TO. On the other hand, immunostaining
`for Bax (apoptosis gene) were mild to moderately positive in the xenografts of
`Groups T12h (12 hours PTx), T24h (hours). T48h (hours) and 72h (hours) but
`negative at Groups TO and T60m (minutes), indicating the involvement of
`programmed cell death in xenograft during pnmary rejection. To evaluate whether
`HO-I and BcI expression can protect grafts from antibody injury, we conducted
`antiserum passive transfer experiments in which hamster heart xenografts were
`challenged with anti-hamster sera (0.5 ml 1.V ) at different time points PTx. We
`found that xenografts challenged with antisera at 60 minutes (n=5) and 12 hours
`(n=5) were hyperacutely rejected (10-18 minutes) despite of HO-I and Bcl-2 gene
`expression by the heart grafts. indicating that intragraft expression of these protection
`genes was not effective for protecting the xenograft from humoral injury. We conclude
`that HO-I and Bcl-2 gene expression in hamster hem xenografts in early stage of
`the humoral rejection has minimal protective effect on xenograft survival, 11 may
`simply reflect a stress-related cellular response of the xenograft tissue to various
`stimuli dunng xenograft rejection.
`
`Abstract# 847
`Poster Board #-Session: P234-I1
`EVALUATION OFDEFEREhToGGALGLYCOCONJUGATES FOR USE
`IN XENOTRANSPLANTATATION. Alexander Schwarz,' Ivona Bakaj,l
`Patrick Birch,' Joanna Fesi,' Anna Nepomich,' Cianna Cooper,' Margaret
`A. Velardo,' Aileen Stark,' Lisa E. Diamond,' John S. Logan,' Guerard W.
`Byme.[ 'Nextran, Princeton. NJ.
`Porcine organs, are rapidly rejected after transplantation into pnmate recipients
`due to the presence of pre-existing immunoglobulins that bind to terminal galactose
`al.3 galactose residues (a-Gal) present on porcine glycoproteins and glycolipids.
`These antibodies are responsible for initiating hyperacute rejection (HAR) of porcine
`organs, and under conditions that block HAR. appear to be induced and contribute
`to the process of acute vascular rejection. Currently available immunosuppressive
`reagents have been largely ineffective at controlling the induction of anti-Gal
`antibodies. Non-antigenic hapten polymers have been shown to be effective materials
`for controlling humoral immune responses in various model systems. As a
`preliminary step toward developing an a-Gal containing polymer to control the
`induced a-Gal antibody we have developed a senes of a-Gal glycoconjugates and
`tested their ability to block anti-Gal antibody binding in vitro. A galactose a1.3
`galactose p 1.4 GlcNAc trisaccharide (TRFA) with a 5 carbon spacer arm was
`coupled to a variety of polymeric backbones including dextran. branched
`polyethylene glycol (PEG). polymethacrylamide (PMA). and poly-L-lysine. The
`ability of these a-Gal conjugates to block anti-Gal IgG and IgM binding was
`determined relative to TRFA in an ELlSA assay using a defined HSA-Gal
`glycoconjugate substrate. The ability of these conjugates to block complement
`dependent cytotoxicity was also determined. When the TRFA sugar was coupled
`to a branched amino PEG carrier using I-ethyl-3(3-dimethylaminopropyl)
`carbodiimide ( E X ) . a uniform product with 8 a-Gal residues was obtained. This
`material was only marginally more effective than TRFA in blocking anti-Gal antibody
`interactions. In contrast a polymer of the branched 8 arm PEG and linear conjugates
`made using Dextran. poly-L-lysine or polymethacrylamide, all with similar antigen
`densities. were much more effective inhibitors of anti-Gal antibodies and anti-Gal
`dependent cytotoxicity. a-Gal conjugates similar to these may be useful both to
`block anti-Gal interactions In vivo and to Specifically control the induced anti-Gal
`immune response.
`
`Poster Board #-Session: P235-I1
`Abstract# 848
`HUMAN MONOCYTES PLAY AN IMPORTANT ROLE IN ISDIRECT
`ANTIGEN PRESENTATION AND COSTIMULATION TOT CELLS
`DURING HUMAN ANTI-PORCINE IMMUNE RESPONSES. He Xu,
`Douglas K. Tadaki, Patrick J. Blair, Francis Cruzata, David M. Harlan, Allan
`D. Kirk.
`Unlike peripheral blood mononuclear cells (PBMC), purified T cells proliferate
`poorly in response to pig endothelial cells (PEC) suggesting that monocytes may
`play a key role in xenogeneic T cell responses. We studied the role of monocytes in
`indirect antigen presentation and costimulation to T cells during human anti-pig
`immunoresponses. The CD40 and CD80 expression on monocytes from a human
`PBMC-PEC coculture was evaluated by FACS. Unlike resting monocytes expressing
`only CD86, monocytes from PBMC-PEC cocultures expressed CDBO and CD40
`as early as 24 hours and high level exprersion at 72 hours of coculture. This
`expresrion was dependent on the presence of T cells, as purified monocytes did not
`up-regulate CD40 and CDRO when incubated alone with PEC. Incubation of purified
`T cells with monocytes harverted from monocyte-PEC cocultures showed up-
`regulation of CD40 and CDRO on monocytes suggesting that these monocytes are
`activated in the presence of naive T cells and that PEC interact with monocytes to
`condition them for activation. Monocytes collcctcd from monocyte-PEC coculture
`werc incubated with purified autologous T cells in the presence or absence of anti-
`CDXO or anti-CDR6 monoclonal antibodies to study the role of conditioned
`monocytes on T cell proliferdtion and costimulation Unlike resting monocytes.
`PEC-activated monocytes induced T cell proliferation and this proliferation was
`inhibited by 8 7 blockade suggesting a key role for B7/CD28 interactions between
`PEC-activated monocytes and T cells i n xenograft rejection. In summary. human
`monocytes exposed to PEC are conditioned to up-regulate costimulatory molecules
`upon expose to T cells. PEC-conditioned monocytes induce T cell proliferation by
`indirect presentation. and costimulatory blockade against B7 inhibits T cell
`proliferation. Our findings suggest that monocytes play a key role in indirect
`antigen presentation and in providing costimulation to T cells through B7/CD28
`pathway. This interaction can occur distant from the initial rile of xenoantigen but
`monocytes remain void of costimulatory signals until interaction with T cells. This
`pathway suggests that human, not porcine. costimulatory molecules are critical
`during xenografts. and demonstrates a mechanism hy which monocyter traffic to
`secondary lymphoid tissue without providing costimulatory signals in the peripheral
`circulation
`
`

`

`Poster Board #-Session: P236-I1
`Abstract# 849
`DEVELOPMENT AND CHARACI'ERIZATION OF ANTI-GAL B-CELL
`RECEPTOR TRANSGENIC GAL-/- MICE. Hui Xu, Ymg Lei, Ajay Sharma,
`Hua Wan, Jeaninne Okabe, John S. Logan, Guerard W. Byme. 'Nextran Inc.
`Princeton, NJ.
`The successful clinical application of xenotransplantation using organs derived
`from pigs IS currently limited by the development of acute vascular rejection a
`process that is thought to involve an induced humoral immune response, largely
`directed towards the a-Gal antigen. Successful transplantation of pig organs into
`humans may require the development of methods for removal or neutralization of
`anti-Gal antibodies and anti-Gal producing B-cells. The large diversity of the B cell
`repertoire makes it difficult however to study anti-Gal B cells. To address this issue
`we have established a transgenic mouse model (BCR;Gal-/-) for investigating anti-
`Gal B cells by introducing a transgene encoding both heavy and light chains for an
`anti-Gal IgM antibody into Gal-/- mice. ELISA analysis of serum from BCR;Gal-/
`
`-. BCR;Gal+/- transgenic mice and Gal-/- mice demonstrates elevated expression of
`
`anti-Gal antibodies in BCR;Gal-I- mice compared to nontransgenic Gal-/- animals.
`Anti-Gal antibody expression in BCR;Gal-/- mice could be increased by
`immunization with an ovalbumin-Gal glycoconjugate in vivo and through stimulation
`with LPS in vitro indicating the presence of functional anti-Gal B cells. Multiparameter
`flow cytometric analysis indicates that 7040% of splenic and peritoneal B cells
`excluded endogenous immunoglobulin gene rearrangement and expressed the
`transgene. FACS analysis of the BCR;Gal-/- B cells identifies them as a population
`of CDS-CD23+ conventional B cells. The majority of these B cells expressed anti-
`Gal receptors on the surface, as identified by FITC-Gal-BSA staining. These
`observations suggest that this model can be used to establish a reliable source of
`anti-Gal B cells which potentially could be used to test the effectiveness of
`immunosuppressive reagents.
`
`Poster Board #-Session: P237-11
`Abstract# 850
`ANTI-GAL ANTIBODY GENE USAGE IN NAIVE AND POST
`XENOCRAFI'GAL-/- MICE. Hui Xu,' Ajay Sharma,' Ltblng Chen,' Caren
`Harrison,' Yuanyuan Wei,' Anita S.-F. Chong,z John S. Logan,' Guerard W.
`Byme.' 'Nextran Inc, Princeton. NJ.
`Naturally occurring antibodies that bind to terminal galactose al.3-galactose
`carbohydrate structures (Gal) are present in humans and Old World Monkeys but
`are negatively regulated in other mammalian species since they express Gal cpitopes
`on their cell wrfaces. A Gal knockout m o w (Gal-/-) model. generated by homologous
`disruption of the a1 ,3-galactosyltransferase gene, is capable of producing natural
`anti-Gal antibodies. To study the genetic control of the anti-Gal response, we have
`generated anti-Gal hybridomas from Gal-/- mice and analyzed the VH genes of
`these antibodies from nayve animals and from mice stimulated by rat heterotopic
`heart transplantation. Six IgM anti-Gal hybridomas derived from naive Gal-/- mice
`exhibited anti-Gal binding activity with some cross reactivity to related carbohydrate
`structures. These naive anti-Gal antibodies used five different VH genes in a germline
`configuration. Anti-Gal IgM hybridomas isolated after a rat heterotopic heart
`xenograft (4 and 21 days) utilized germline VH gene segments from the VH7183
`family and exhibited less cross reactivity. In contrast, we have predominantly
`isolated IgG anti-Gal hybridomas from mice 21-days after rat heterotopic heart
`xenografts, indicating an isotype switch. Nine of the IgG anti-Gal hybridomas
`secreted the lgG3 subclass and one produced IgGI. Sequence analysis of the VH
`gene usage from the induced anti-Gal IgG antibodies demonstrated a restncted gene
`utilization. VHJ606-14A. Our results demonstrate that the anti-Gal response in
`nai've Gal-/- mice i* encoded hy multiple germline progenitors. In response to a
`xenograft. the induced anti-Gal antibodies exhibited a restncted gene usage and
`somatic mutations. indicating a positive selection
`
`Poster Board #-Session: P238-I1
`Abstract# 851
`ANTI-GAL MONOCLONAL ANTIBODIES INDUCE PIG
`MICROVASCULAR E"IlLELIALCELL(PMVEOACIWATI0NAND
`APOPTOSIS. Hui Xu. Bashoo Naziruddin, Yuanyuan Wei, Libing Chen,
`John S. Logan, Guerard W. Byme. 'Nextran Inr. Princeton, NJ
`We have produced a series of anti-Gal hybndomas from Gal -/- mice after rat to
`mouse heterotopic cardiac xenografts. These monoclonal anti-Gal antibodies provide
`us an opportunity to study the effects that anti-Gal antibodies may have on
`endothelial cells in a very defined manner. All IgG3 anti-Gal monoclonal antibodies
`bound to pig red blood cells (RBC). and induced complement dependent lysis. In
`contrast. an lgGl anti-Gal antibody tailed to activate complement. Incubation of
`anti-Gal monoclonal antibodies with cultured porcine microvascular endothelial
`cells (PMVEC) for 2-4 hours resulted in activation of endothelial cells as determined
`by the induction of E-selectin (CD62E). This activation was anti-Gal specific since
`the induction of CD62E by anti-Gal hybridomas was blocked by IOmM free a-Gal
`trisacharride and since a mouse IgG isotype control failed to induce CD62E
`expression. When antibodies were incubated with endothelial cells overnight at hlgh
`concentration. anti-Gal antibodies also induced apoptosis as determined by Annexin
`V staining. The induction of apoptosis was dose dependent within the range of S-
`SO pg/ml and isotype controls. mouse lgC3 and IgGl. did not induce significant
`apoptosis. The induction of endothelial cell activation. or apoptosis showed no
`
`XENOTRANSPLANTATION II
`
`apparent relationship to the affinity of the anti-Gal antibody. These data suggest
`that induced anti-Gal antibodies are capable of effecting the endothelial cells of a
`graft and could induce the formation of a procoagulant vasculature, without the
`need of complement mediated damage.
`
`Poster Board #-Session: P239-I1
`Abstract# 852
`PIG-TO-PRIMATE RENAL XENOTRANSPLANTATION -
`PREDOMINANT GLOMERULAR INJURY IN LONG TERM
`FUNCTIONING H-CD55 TRANSGENIC KIDNEYS. Jan Schmidtko,'
`Martin Loss.' Ergin Kilic,? Ralf Roland Lorenz,' Jens Martin Hecker,' Richard
`Appiah,' Robert Kun2.l Juergen Klempnauer,' Udo Helmchen,2 Michael
`Winkler. I 'Klinik fur Viszeral- rind Transplantationschrriir~ie, Medizinisrhe
`Hochschule Hannover, Hannover. Germany; 'Institrct fiir Pathologie.
`Universitaets-Klinikum Eppendorf, Hamburg, Germany; 'Zentrum
`Anaesthesiologie, Medizinische Hochschule Hannover, Hannover.
`Germany.
`The introduction of transgenic pigs expressing human complement regulatory
`molecules such as h-CD5S or h-CDS9 into pig to primate discordant kidney
`xenotransplantation has lead to xenograft survival times consistently above 1 week.
`This allows for a more refined analysis of discordant xenograft pathology during
`longer graft survival times. Methods: H-CDSS positive porcine kidneys were
`transplanted in cynomolgus monkeys under postoperative cyclophosphamide,
`cyclosporine A and steroid baseline immunosuppression and CI -1nh treatment for
`episodes of acute vascular rejection (AVR). We here report on a subgroup of
`experiments ( n 4 ) with graft survival times of more than 2 weeks. In all experiments
`graft biopsies were obtained at regular intervals. Histological monitoring was
`performed by means of immunohistochemistry (FVIII-RAg, IgM, IgG, C3, CSb-9.
`h-CDSS). standard PAS staining and transmission electron microscopy (TEM).
`Results: The median survival time in this cohort of 4 animals was 24.5 days,
`longest survival achieved was 68 days. In all biopsies investigated. despite seemingly
`stable graft function a progressive graft damage was observed. predominantly on
`the glomerular level. TEM analysis revealed profound glomerular basement
`membrane damage over time. On immunohistochemistry this damage was paralleled
`by an increased deposition of recipient IgM immunoglobulins associated with
`subsequent deposition of terminal complement complexes. Discussion: In this pig
`to primate kidney transplantation model. despite profound postoperative CyP
`based immunosuppression and seemingly stable graft function for > two weeks, a
`subacute graft damage likely to be mediated by intragraft recipient antiporcine
`antibody deposition is induced by the recipients humoral immune system.
`
`Poster Board #-Session: P240-I1
`Abstract# 853
`NATURAL REGULATION OFGALUl-3CAL EXPRESSION. JOO HoTai,'
`Jeffrey L. Platt.'.','
`'Surgery. Mayo Clinic, Rochester, MN: 'Immunology.
`Mayo Clinic. Rochester. MN; 'Pediatrics. Mayo Clinic. Rochester, MN.
`One primary hurdle to clinical xenotransplantation IS expression of a sacchande,
`Galal-3Gal. by lower mammals such as pigs. This saccharide is recognized by
`xenoreactive antibodies which initiate the vascular rejection of pig-to-primate
`xenografts. The expression of Galal-3Gal is presumably controlled by the
`availability of core saccharide precursor, nucleotide sugars and the reciprocal actions
`of a- I .3-galactosyltransferase and a-galactosidase. To elucidate the mechanisms
`controlling expression of Galal-3Gal. we studied porcine testis. a tissue which
`exhibits a dramatic transition of Galal -3Gal expression. using lectins, and antibodies
`
`directed against a-I .3-galaciosyltr~~~erase and a-galactosidase. To localize porcine
`a-galactosidase as well as porcine a- I ,3-galactosyltransferase. we cloned the
`corrcsponding genes by 5'/3' RACE (rapid amplification of cDNA ends) and
`developed antibodies against deduced amino acid sequences. Primary spermatocytes,
`but not more mature spermatocytes bound the 1B4 lectin. indicating expression of
`Galal -3Gal. Spermatogonia bound UEA-I and secondary spermatocytes and some
`of spermatids bound LEL and RCA-I lectins instead of 184. suggesting loss of
`terminal a-galactosyl residues or de noro synthesis of core structure with maturation
`toward the center of seminiferous tubules. Munne monoclonal antibodies specific
`for porcine bound to senoli cells and spermatozoa in spcrmiation. apparently in
`middle pieces of tail. while monoclonal antibodies specific for porcine a-galactosidase
`bound 10 sertoli cells and the region including secondary spermatocytes, spermatids
`and residual bodies. These results suggest that the dramatic transition of Galal-
`3Gal expression in seminiferous tubules might be caused by the activity of a-
`galactosidase and/or possibly by the cleavage of acceptor for a-1.3-
`galactosyltrdnsfere. This observation offers a potential means by which Galal -
`3Gal expression might be modulated.
`
`349
`
`

`

`ANTIGEN PRESENTATION
`
`Poster Board #-Session: P241-I1
`Abstract# 854
`PERIPHERAL SURVIVAL OF MURINE C M T CELLSSELECTED IN
`PORCINE THYMUS CRAIWS. Jose-Ignacio Rdriguez-Barbosa,l Yong
`Zhao,' Gui-Ling Zhao,' Sheng-Ping Wang,' David H. Sachs,' Megan Sykes.'
`'Bone Marrow Transplantation Section. Transplantation Biology
`Research Center, Massachusetts General HospitallHarvard Medical
`School, Boston, MA.
`Background: We have demonstrated that thymus transplantation IS capable of
`inducing tolerance across xenogeneic barriers. Studies in T cell receptor transgenic
`mice showed that positive selection in porcine thymus grafts is mediated entirely
`by porcine MHC. Some reports suggest that T cells positively selected on a
`piuticular thymic MHC require the same MHC in the penphery to survive. This
`observation may pose a problem for the strategy of using xenogeneic thymic
`transplantation for the induction of tolerance. since T cells are selected on an MHC
`different from that they will see in the periphery. Methods: We compared the
`decay of mouse CD4 T cells selected in fetal porcine thymus (FPTHY) grafts with
`that of T cells selected in syngeneic neonatal mouse thymus (NMTHY) grafts
`following removal of
`the thymic graft from thymectomized (ATX) conditioned
`class 11 wild type (WT) and class 11 knock-out (KO) mice. Munne CD4 repopulation
`was followed by FACS. These studies assessed whether continual CD4 cell
`repopulation in conditioned FPTHY WT mice requires continual output of T cellc
`from porcine thymic grafts to compensate for rapid death of T cells . Results :
`Conditioned ATX class 11 WT and class II KO mice grafted with either FPTHY
`or syngeneic NMTHY reconstituted the penpheral CD4 T cell compartment to the
`same extent. Following graftectomy at 13 weeks. a gradual decline was observed
`among CD4' PBL and naive-type CD4 cells in class 11 WT and class II KO mice
`grafted with NMTHY. Only at 15 weeks after Gra!Tx was it possible to detect a
`statistically significant increase in the decay of CD4 cells in GrafTx class I1 KO
`compared to WT controls (p4.05). The decline of the % C W PBL and naive-
`type CD4 (CD4+CD44"w) cells after GrafTx was also gradual and similar in class
`11 WT mice grafted with either NMTHY or FFTHY. Conelusion : A munne CD4
`T cell repertoire positively selected on porcine thymus is able to survive in the
`periphery of mice, despite the apparent absence of porcine antigen presenting
`cells. The same class I1 in the periphery as that of the thymus in which T cells were
`selected may not play a cntical role in the survival of T cells selected on a highly
`disparate xenogeneic MHC.
`
`Abstract# 855
`Poster Board #-Session: P242-I1
`ROLE OF PIG CYTOKINES IN INDUCTION OF XENOGENEIC BONE
`MARROW CHIMERISM IN A DISCORDAWPIGTO MOUSE MODEL
`Muhammad M. Mohiuddin,! Yuru Meng,' Verdi J. DiSesa.' 'Cardiovascular-
`Thoracic Surgery, Rush Presbyterian St Luke's Medical Center, Chicago,
`IL.
`Purpose :Xenografts from pigs might solve the problem of the shortage of donor
`organs. The immune mechanisms of xenograft rejection are complex and cannot be
`controlled by conventional immunosuppressive drugs. Induction of donor specific
`tolerance would over come this bamer. Mixed xenogeneic chimerism might be an
`effective method of inducing tolerance to xenografts. In this study we demonstrate
`the importance of pig cytokines in long term survival of pig bone marrow cellc in a-
`Gal knockout (KO) and CS7BL6 (WT) mice. Methods : Pig hone marrow was
`extracted from nbs and a single cell suspension of whole bone marrow ( 60 -100 X
`100celIs) was injected into sub-lethally irradiated (6SOcCy) a-Gal KO (Group A)
`and WT (Group B) mice(n=lO in each group). Both groups of mice received 0.1 ml
`of Anti Lymphocyte Serum (ALS) on day -1 and a single dose(SOmg/Kg) of
`Cyclophosphamide (CyP) on day 2. Half of the mice from each group received Pig
`IL3 and GM-CSF (IOpgKg) daily. All mice were typed by flow cytometry every
`4 weeks using Lectin-FITC and SLA-DR antibodies. Results: At four weeks Group
`A mice which received cytokines showed significani multi-lineage engraftment of
`pig cells (6.8%) whereas, mice not receiving cytokines \howed no engraftment of
`pig cells (0%). In group B mice which received cytokines, a very small number of
`pig cells were detected (2.3%) whereas, in mice whichdid not received any cytokine\.
`a high number of pig cells was detected (12-14%). At eight week\ only group A
`mice receiving cytokines and group B mice not receiving cytokines had permtcnce
`of multilineage pig cells (2-6%). No pig cells were dctccted at eight weeks in the
`other two groups. Conclusion: We have demonstrated that stable engraftment of
`pig cells in KO mice is dependent on pig cytokines whereas in wild type mice pig
`cytokines are not required. Perhaps, in KO mice cytokine producing cells in the pig
`bone marrow inoculum are suppressed or killed by the anti a-Gal antibodies whereas.
`in WT mice this bamer is absent. This study also indicates that the WT mou\e
`hematopoietic environment is sufficient for the growth of pig bone marrow cells.
`
`Abstract# 856
`Poster Board #-Session: P243-I1
`ACllVA"IONOFHUMANDENDRITICCEL,LSBY PORCINEAORTIC
`ENDOTHELIAL CELLS: TRANSACTIVATION OF NAIVE T CELLS
`THROUGH C(ISIIMULATIONANDCYTOKINEGENERATI0N. Partha
`P. Manna, T. Mohanakumar.
`Background: Debdritic cells (DCs) are mosi potent antigen presenting cells in the
`immune system. In order 10 define the role of human DC in human anti-porcine
`immune responses. we analyzed the interaction of human DC with porcine aortic
`endothelial cells(PAEC).
`Methods, To determine the immune responses, both monocyte derived and
`peripheral blood D C s was cultured with porcine and human endothelial cells. We
`analyzed the role of CDI la, CDI I band CDS4 in cell to cell adhesion assay using
`antibodies against these molecules. The expression pattern of co-stimulatory
`molecules (CD40,CDXO,CD86),adhesion molecule (CDS4), and intracellular
`cytokines (IL-12~70 and TNF-U) in DC following interaction with endothelial
`cells was determined by immunofluorescence.
`Results: Human DC adhered to PAEC (3R-402) and this adhesion was augmented
`(>SO%) upon treatment with either recombinant swine interferon-y or recombinant
`human TNF-a. Addition of human DC to PAEC was blocked by pretreatment of
`DC with antibodies specific to human LFA-I and CDS4. Adhesion of DC to PAEC
`also resulted in the activation of DC which was manifested by upregulation of co-
`stimulatory molecules (CLMO,CDXO,CDX6), adhesion molecule (CD54) and HLA-
`DR. PAEC activated human DC provided proliferative signals to the naive autologous
`CD4+ T cells and synthesized IL-12~70 and TNF-a. However, activated DCs
`failed to lyse PAEC in such interaction.
`Conclusion: Human DCs effectively adhered to PAEC and are activated by
`xenoantigen resulting in highly efficient antigen presentation and proliferation of
`CD4+ T cells. Further, this interaction of human DC to PAEC is regulated by the
`participation of co-stimulaiory molecules (CD40.CDRO.CD86). adhesion molecules
`(LFA-I.CDS4) and cytokines (IL-12p70,TNF-a).
`
`CONCURRENT SESSION 41 :
`h G E N PRESENTATION
`
`Abstract# 857
`Uh"PRIMEDCD&TCELLSAREACI'IVATEDBY RESIINGVASCULAR
`ENDOTHELIUM TO INDUCE ENDOTHELIAL CELL APOPTOSIS.
`Alexander S. Krupnick,l Daniel Kreisel.' Wilson Y. Szeto,' Sicco H. P0pma.l
`Bruce R. Rosengard.' 'Department nf Surgery. Hospital of the University of
`Pennsylvania, Philadelphia, PA.
`Purpose: Immunologic destruction of endothelium contributes to acute rejection as
`well as chronic allograft vasculopathy. Although activated vascular endothelium
`has been classified as a semiprofessional antigen presenting cell, it IS controversial
`whether resting vascular endothelium present at the blood/allograft interface can
`activate allogeneic T cells without help from profesbional antigen presenting cells
`(APC). Our purpose is to define this interaction in an in vitro system.
`Methods: Vascular endothelial cells from the thoracic aorta of CS7BW6 (H-2Kh)
`mice were isolated and cultured free of professional APC using a recently developed
`method. CD8+ T lymphocytes were punfied from splenocytes and pooled lymph
`node cells of TCR transgenic (reactive against H-2Kh) BM3(H2KL) mice using
`nylon wool depletion of adherent cells and positive selection with immunomagnetic
`beads. The CDX+ T cells were labeled with CFSE and proliferation was tracked
`using flow cytometry on day\ 2.4. and 6 Apoptosis of the endothelium was
`evaluated concurrently by TUNEL staining.
`Rewlts. Only minimal T cell proliferation ((1.5% and 3%)was seen on days 2 and 4
`respectively. but SS% of the T cells had proliferated by day 6. Accordingly only
`0.5% and 16% of the endoihelial targets were destroyed by ihe alloreactive T cells
`on days 2 and 4. while 41% of the endothelium was undergoing apopto\is 6 days
`after co-culture compared to control\.
`Conclusion: We have demonstrated that mouse vascular endothelium can activate
`unpnmed alloreactive CDX+ T lymphocytes which then induce endothelial cell
`apoptosis. These results sugge\t that resting vascular endothelium can act as an
`antigen presenting cell for CDX+ direct allorecognition and may therefore play an
`important role in regulating immune processes of acute and chronic allograft rejection.
`Since endothelium remains at the blWallograft interface for the life of the patient,
`these finding have important immunological consequences for the field of organ
`transplantation.
`
`350
`
`

`

`Abstract# 858
`INTERACTIONS AMONG RECIPIENT MONOCYTES AND DONOR
`I N T H E M A I " A N C E 0 F T H E I P A T H W A Y
`OF ALLORECOGNITION. Dmitry V. Samsonov,' Christopher S. Geehan,'
`Mark D. Denton,' Gilles Benichou,2 Ana Maria Waaga,l Mohamed H.
`Sayegh,' David M. Briscoe.' 'Division ofNephrology. Children's Hospital,
`Boston, MA; ZSchepens Eye Research Institute, Harvard Medical School,
`Boston, MA.
`Endothelial cells (EC) promote the differentiation of monocytes into immature
`APCs. These immature APCs are primed to phagocytose apoptotic and necrotic
`cells within the graft. We have shown that monocyte-endothelial interactions that
`promote APC generation also induce the production of proinflammatory cytokines
`by the APC. We now propose that this function of EC facilitates the maintenance
`of Tcell activation via the indirect pathway of allorecognition. To test this possibility,
`we first cultured PBMCs with allogeneic EC for 6 days. CD14+ monocytes were
`isolated from the coculture and were used as stimulators with autologous CD4+ T
`cells. In these studies, the proliferation of T cells was 3-6 times higher in the
`presence of these EC-modified monocytes as compared to unmodified monocytes
`cultured on plastic. Thus, EC either enhance the costimulatory capacity of APCs.
`or/and promote proliferation via the indirect pathway of allorecognition. To assess
`the latter possibility, we compared proliferation of CD4+ T cells to the direct or the
`indirect pathway when EC were used as a source of alloantigen. CD4+ T cells were
`cultured with irradiated self-APCs in the presence of either allogeneic EC monolayers
`or sonicated EC. T cell proliferati

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket